Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMJ Case Rep ; 14(11)2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34799385

RESUMEN

Soft tissue sarcomas (STSs) are rare and may often be misdiagnosed, resulting in delays in treatment. A 67-year-old cisgender woman with type 2 diabetes mellitus and obesity presented to her primary care physician with a mass on her left proximal arm. The clinical opinion of the attending physician was that of an insulin injection site reaction. After further evaluation from the physician, the patient was diagnosed with a lipoma without confirmatory histology. The patient continued to present with an enlarging mass, decline in health status and continued with local wound care. The patient underwent a confirmatory biopsy following which, the patient was diagnosed with leiomyosarcoma. This case report highlights the case of a person with a low or moderate income with a self-reported low health literacy living in a rural community and how STS may be misdiagnosed in medically underserved. The patient's primary or oncology care team are not involved in the production or review of this case report.


Asunto(s)
Diabetes Mellitus Tipo 2 , Leiomiosarcoma , Neoplasias Cutáneas , Anciano , Femenino , Humanos , Reacción en el Punto de Inyección , Insulina , Leiomiosarcoma/diagnóstico
2.
Clin Lab Med ; 38(1): 67-90, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29412886

RESUMEN

The process of conducting pathology research in Africa can be challenging. But the rewards in terms of knowledge gained, quality of collaborations, and impact on communities affected by infectious disease and cancer are great. This report reviews 3 different research efforts: fatal malaria in Malawi, mucosal immunity to HIV in South Africa, and cancer research in Uganda. What unifies them is the use of pathology-based approaches to answer vital questions, such as physiology, pathogenesis, predictors of clinical course, and diagnostic testing schemes.


Asunto(s)
Investigación Biomédica , Servicios de Laboratorio Clínico , Patología Clínica , África , Países en Desarrollo , Humanos
3.
Clin Cancer Res ; 23(12): 3061-3071, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-27852699

RESUMEN

Purpose: This study examines cell surface ROR1 expression in human tumors and normal tissues. ROR1 is considered a promising target for cancer therapy due to putative tumor-specific expression, and multiple groups are developing antibodies and/or chimeric antigen receptor-modified T cells to target ROR1. On-target, off-tumor toxicity is a challenge for most nonmutated tumor antigens; however, prior studies suggest that ROR1 is absent on most normal tissues.Experimental Design: Our studies show that published antibodies lack sensitivity to detect endogenous levels of cell surface ROR1 by immunohistochemistry (IHC) in formalin-fixed, paraffin-embedded tissues. We developed a ROR1-specific monoclonal antibody (mAb) targeting the carboxy-terminus of ROR1 and evaluated its specificity and sensitivity in IHC.Results: The 6D4 mAb is a sensitive and specific reagent to detect cell surface ROR1 by IHC. The data show that ROR1 is homogenously expressed on a subset of ovarian cancer, triple-negative breast cancer, and lung adenocarcinomas. Contrary to previous findings, we found ROR1 is expressed on several normal tissues, including parathyroid; pancreatic islets; and regions of the esophagus, stomach, and duodenum. The 6D4 mAb recognizes rhesus ROR1, and ROR1 expression was similar in human and macaque tissues, suggesting that the macaque is a suitable model to evaluate safety of ROR1-targeted therapies.Conclusions: ROR1 is a promising immunotherapeutic target in many epithelial tumors; however, high cell surface ROR1 expression in multiple normal tissues raises concerns for on-target off-tumor toxicities. Clinical translation of ROR1-targeted therapies warrants careful monitoring of toxicities to normal organs and may require strategies to ensure patient safety. Clin Cancer Res; 23(12); 3061-71. ©2016 AACR.


Asunto(s)
Carcinoma/tratamiento farmacológico , Carcinoma/genética , Inmunoterapia , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Anticuerpos Monoclonales/inmunología , Carcinoma/inmunología , Carcinoma/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Terapia Molecular Dirigida , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/inmunología , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/aislamiento & purificación , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
4.
Proc Natl Acad Sci U S A ; 113(31): 8795-800, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27439864

RESUMEN

Insulin receptor substrate-1 (IRS-1) is a signaling adaptor protein that interfaces with many pathways activated in lung cancer. It has been assumed that IRS-1 promotes tumor growth through its ability to activate PI3K signaling downstream of the insulin-like growth factor receptor. Surprisingly, tumors with reduced IRS-1 staining in a human lung adenocarcinoma tissue microarray displayed a significant survival disadvantage, especially within the Kirsten rat sarcoma viral oncogene homolog (KRAS) mutant subgroup. Accordingly, adenoviral Cre recombinase (AdCre)-treated LSL-Kras/Irs-1(fl/fl) (Kras/Irs-1(-/-)) mice displayed increased tumor burden and mortality compared with controls. Mechanistically, IRS-1 deficiency promotes Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling via the IL-22 receptor, resulting in enhanced tumor-promoting inflammation. Treatment of Kras/Irs-1(+/+) and Kras/Irs-1(-/-) mice with JAK inhibitors significantly reduced tumor burden, most notably in the IRS-1-deficient group.


Asunto(s)
Adenocarcinoma/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células A549 , Adenocarcinoma/genética , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Femenino , Humanos , Proteínas Sustrato del Receptor de Insulina/deficiencia , Proteínas Sustrato del Receptor de Insulina/genética , Estimación de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones Noqueados , Persona de Mediana Edad , Mutación , Fenotipo , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Transducción de Señal/genética
5.
Transplant Direct ; 1(2)2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25988188

RESUMEN

BACKGROUND: CD28 signal blockade following T cell receptor activation is under intense investigation as a tolerance-inducing therapy for transplantation. Our goal is to produce a CD28-specific reagent as a therapy for the prevention of graft rejection and graft-versus-host disease in the canine model of allogeneic hematopoietic cell transplantation (HCT). METHODS: We infused a monoclonal mouse anti-canine CD28 antibody (1C6 mAb) into four dogs and a fragment of antigen-binding (1C6 Fab) into two dogs. Pharmacokinetics, pathology, cytokine release, and the crystal structure of 1C6 Fv were evaluated. RESULTS: Within an hour of an IV injection of the 1C6 mAb, the dogs became leukopenic and developed a steroid-refractory cytokine storm. Two of the dogs developed high fevers, one experienced diffuse alveolar hemorrhage, and another developed gastrointestinal hemorrhage. The cytokine storm was characterized by elevated plasma levels of MCP-1, IP-10, IL-10, IL-6, and TNF-α. In addition, one dog showed elevated levels of IL-2, IL-8, and IL-18. In contrast, infusion of 1C6 Fab was well tolerated without any side effects. Dry-coating 1C6 mAb onto tissue culture plates induced CD3-independent proliferation and TNF-alpha production. Crystal structure analysis revealed that 1C6 binds to canine CD28 in a manner different than previously reported for conventional agonistic or superagonistic antibodies. CONCLUSIONS: These results indicate that dogs and humans develop a similar cytokine storm following infusion ofanti-CD28 mAb, providing an appropriate large animal for further study. 1C6 Fab warrants evaluation as a tolerance-inducing reagent in the canine model of allogeneic HCT.

6.
J Histochem Cytochem ; 62(3): 197-204, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24152994

RESUMEN

Immunohistochemistry on mouse tissue utilizing mouse monoclonal antibodies presents a challenge. Secondary antibodies directed against the mouse monoclonal primary antibody of interest will also detect endogenous mouse immunoglobulin in the tissue. This can lead to significant spurious staining. Therefore, a "mouse-on-mouse" staining strategy is needed to yield credible data. This paper presents a method that is easy to use and highly flexible to accommodate both an avidin-biotin detection system as well as a biotin-free polymer detection system. The mouse primary antibody is first combined with an Fab fragment of an anti-mouse antibody in a tube and allowed sufficient time to form an antibody complex. Any non-complexed secondary antibody is bound up with mouse serum. The mixture is then applied to the tissue. The flexibility of this method is confirmed with the use of different anti-mouse antibodies followed by a variety of detection reagents. These techniques can be used for immunohistochemistry (IHC), immunofluorescence (IF), as well as staining with multiple primary antibodies. This method has also been adapted to other models, such as using human antibodies on human tissue and using multiple rabbit antibodies in dual immunofluorescence.


Asunto(s)
Anticuerpos/inmunología , Inmunohistoquímica/métodos , Animales , Anticuerpos/química , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Biotina , Técnica del Anticuerpo Fluorescente , Cabras , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/inmunología , Indicadores y Reactivos , Ratones , Especificidad de Órganos , Conejos , Coloración y Etiquetado/métodos
7.
Vet Dermatol ; 24(3): 346-54, e76-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23600606

RESUMEN

OBJECTIVES: To describe the clinical, histopathological and immunohistochemical lesions and the response to therapy of a novel skin disease in four dogs, and to compare the lesions with those of other similar conditions. METHODS: Clinical lesions, the histopathological findings in skin biopsy samples, immunohistochemistry for CD3 and cleaved caspase-3 and the response to therapy were evaluated. RESULTS: Clinical lesions included multifocal, coalescing, verrucous, crusted papules and plaques with erythematous borders and comedones or follicular casts. Lesions were in haired skin; they occurred at the edges of paw pads and claw beds in one dog. Histopathological lesions included ortho- and more prominent parakeratotic hyperkeratosis involving follicular infundibular epithelium, with cast formation and a papillary epidermal surface. Lymphocytic exocytosis affected all strata of follicular infundibular epithelium and epidermis. Variable numbers of acidophilic shrunken keratinocytes, often bordered by lymphocytes (satellitosis), occupied the more superficial strata of the follicular infundibular epithelium and epidermis. Immunohistochemistry revealed numerous CD3+ T lymphocytes and fewer cleaved caspase-3-positive apoptotic keratinocytes in the infundibular hair follicle epithelium and epidermis, with numerous CD3+ T lymphocytes and cleaved caspase-3-positive cells in the dermis. Two dogs responded completely to therapy with ciclosporin and remained lesion free off therapy; one dog responded to therapy with prednisone, azathioprine and ciclosporin, but relapsed; and one dog was not treated. CONCLUSIONS AND CLINICAL IMPORTANCE: The cause of the lesions is unknown; the presence of intraepithelial CD3+ lymphocytes and cleaved caspase-3-positive apoptotic keratinocytes and the positive response to immunosuppressive therapy suggest an immune response directed towards unidentified antigens expressed on the surface of keratinocytes.


Asunto(s)
Apoptosis/fisiología , Dermatitis/veterinaria , Enfermedades de los Perros/patología , Foliculitis/veterinaria , Inmunosupresores/uso terapéutico , Animales , Dermatitis/tratamiento farmacológico , Dermatitis/patología , Enfermedades de los Perros/tratamiento farmacológico , Perros , Femenino , Foliculitis/tratamiento farmacológico , Foliculitis/patología
8.
Proc Natl Acad Sci U S A ; 109(20): 7859-64, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22550175

RESUMEN

The Sonic Hedgehog (Shh) pathway drives a subset of medulloblastomas, a malignant neuroectodermal brain cancer, and other cancers. Small-molecule Shh pathway inhibitors have induced tumor regression in mice and patients with medulloblastoma; however, drug resistance rapidly emerges, in some cases via de novo mutation of the drug target. Here we assess the response and resistance mechanisms to the natural product derivative saridegib in an aggressive Shh-driven mouse medulloblastoma model. In this model, saridegib treatment induced tumor reduction and significantly prolonged survival. Furthermore, the effect of saridegib on tumor-initiating capacity was demonstrated by reduced tumor incidence, slower growth, and spontaneous tumor regression that occurred in allografts generated from previously treated autochthonous medulloblastomas compared with those from untreated donors. Saridegib, a known P-glycoprotein (Pgp) substrate, induced Pgp activity in treated tumors, which likely contributed to emergence of drug resistance. Unlike other Smoothened (Smo) inhibitors, the drug resistance was neither mutation-dependent nor Gli2 amplification-dependent, and saridegib was found to be active in cells with the D473H point mutation that rendered them resistant to another Smo inhibitor, GDC-0449. The fivefold increase in lifespan in mice treated with saridegib as a single agent compares favorably with both targeted and cytotoxic therapies. The absence of genetic mutations that confer resistance distinguishes saridegib from other Smo inhibitors.


Asunto(s)
Meduloblastoma/tratamiento farmacológico , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Alcaloides de Veratrum/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Secuencia de Bases , Western Blotting , Hibridación Genómica Comparativa , Cartilla de ADN/genética , Resistencia a Antineoplásicos , Citometría de Flujo , Perfilación de la Expresión Génica , Inmunohistoquímica , Factores de Transcripción de Tipo Kruppel/genética , Imagen por Resonancia Magnética , Meduloblastoma/patología , Ratones , Datos de Secuencia Molecular , Proyectos Piloto , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Receptor Smoothened , Análisis de Supervivencia , Alcaloides de Veratrum/uso terapéutico , Proteína Gli2 con Dedos de Zinc
9.
Cell Cycle ; 7(13): 2056-70, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18642446

RESUMEN

A balance between angiogenesis inducers and inhibitors in the microenvironment controls the rate of new blood vessel formation. We hypothesized that fibroblasts, an important cellular constituent of the tissue stroma, secrete molecules that contribute to this balance. We further hypothesized that fibroblasts secrete molecules that promote angiogenesis when they are in a proliferative state and molecules that inhibit angiogenesis when they are not actively cycling (quiescent). Microarray analysis revealed that angiogenesis inducers and inhibitors are regulated as fibroblasts transition into a quiescent state and reenter the cell cycle in response to changes in serum. To assess whether changes in transcript levels result in changes in the levels of secreted proteins, we collected conditioned medium from proliferating and quiescent fibroblasts and performed immunoblotting for selected proteins. Secreted protein levels of the angiogenesis inhibitor pigment epithelium derived factor (PEDF) were higher in quiescent than proliferating fibroblasts. Conversely, proliferating fibroblasts secreted increased levels of the angiogenesis inducer vascular endothelial growth factor-C (VEGF-C). For the angiogenesis inhibitor thrombospondin-2, quiescent cells secreted a prominent 160 kDa form in addition to the 200 kDa form secreted by proliferating and restimulated fibroblasts. Using immunohistochemistry we discovered that fibroblasts surround blood vessels and that the angiogenesis inhibitor PEDF is expressed by quiescent fibroblasts in uterine tissue, supporting a role for PEDF in maintaining quiescence of the vasculature. This work takes a new approach to the study of angiogenesis by examining the expression of multiple angiogenesis regulators secreted from a key stromal cell, the fibroblast.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/metabolismo , Proteínas del Ojo/metabolismo , Fibroblastos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Serpinas/metabolismo , Trombospondinas/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Proliferación Celular , Medios de Cultivo Condicionados , Factores de Crecimiento Endotelial/metabolismo , Fibroblastos/citología , Humanos , Neovascularización Fisiológica , Análisis por Matrices de Proteínas , Células del Estroma/metabolismo
10.
J Histochem Cytochem ; 56(4): 347-58, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18071065

RESUMEN

Fibroblasts are critical for tissue homeostasis, and their inappropriate proliferation and activation can result in common and debilitating conditions including fibrosis and cancer. We currently have a poor understanding of the mechanisms that control the growth and activation of fibroblasts in vivo, in part because of a lack of suitable fibroblast markers. We have taken advantage of an antibody previously shown to stain stromal cells in frozen tissues (TE-7) and identified conditions in which it can be used to stain fibroblasts and myofibroblasts in the paraffin-embedded tissue samples routinely collected for pathological analysis. We show that this antibody recognizes growing and quiescent fibroblasts and myofibroblasts by immunohistochemistry, immunofluorescence, and ELISA assays. We also present its staining patterns in normal tissue samples and in breast tumors.


Asunto(s)
Fibroblastos/citología , Especificidad de Anticuerpos , Neoplasias de la Mama/patología , Células del Tejido Conectivo/citología , Vasos Coronarios/citología , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibroblastos/metabolismo , Fijadores , Formaldehído , Humanos , Inmunohistoquímica , Músculo Esquelético/irrigación sanguínea , Adhesión en Parafina , Piel/citología , Células del Estroma/citología , Timo/citología
11.
Infect Immun ; 76(3): 952-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18039832

RESUMEN

Toll-like receptors and the beta-glucan receptor, dectin-1, mediate macrophage inflammatory responses to Aspergillus fumigatus through MyD88-dependent and -independent signaling mechanisms; however, pulmonary inflammatory responses in MyD88-deficient mice challenged with A. fumigatus are poorly defined. The role of MyD88 signaling in early pulmonary inflammation and fungal clearance was evaluated in C57BL/6J wild-type (WT) and MyD88-deficient (MyD88-/-) mice. Early (<48 h) after infection, MyD88-/- mice had higher fungal burdens than those of WT mice, although fungal burdens rapidly declined (>72 h) in both. MyD88-/- mice had less consolidated inflammation, with fewer NK cells, in lung tissue early (24 h) after infection than did WT mice. At the latter time point, MyD88-/- mouse lungs were characterized by a large amount of necrotic cellular debris and fibrin, while WT lungs had organized inflammation. Although there were equivalent numbers of macrophages in WT and MyD88-/- mouse lung tissues, MyD88-/- cells demonstrated delayed uptake of green fluorescent protein-expressing A. fumigatus (GFP-Af293); histologically, MyD88-/- mouse lungs had more hyphal invasion of terminal airways and vessels, the appearance of bronchiolar epithelial cell necrosis, and necrotizing vasculitis. MyD88-/- lung homogenates contained comparatively decreased amounts of interleukin-1beta (IL-1beta), IL-6, KC, and gamma interferon and paradoxically increased amounts of tumor necrosis factor alpha and macrophage inflammatory protein 1alpha. These data indicate that the MyD88-dependent pathway mediates acute pulmonary fungal clearance, inflammation, and tissue injury very early after infection. Resolution of abnormalities within a 3-day window demonstrates the importance of redundant signaling pathways in mediating pulmonary inflammatory responses to fungi.


Asunto(s)
Aspergilosis/inmunología , Aspergilosis/patología , Aspergillus fumigatus/inmunología , Pulmón/inmunología , Pulmón/microbiología , Factor 88 de Diferenciación Mieloide/inmunología , Animales , Aspergilosis/microbiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Recuento de Colonia Microbiana , Citocinas/análisis , Inflamación/patología , Células Asesinas Naturales/inmunología , Pulmón/química , Pulmón/patología , Macrófagos Alveolares/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Necrosis/patología , Neutrófilos/inmunología
12.
Am J Physiol Renal Physiol ; 292(1): F304-12, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16885150

RESUMEN

Acute renal failure (ARF) induces tubular hyperresponsiveness to TLR4 ligands, culminating in exaggerated renal cytokine/chemokine production. However, the fate of TLR4 protein during acute tubular injury remains unknown. The study sought new insights into this issue. Male CD-1 mice were subjected to 1) unilateral ischemia-reperfusion (I/R), 2) cisplatin (CP) nephrotoxicity, or 3) glycerol-induced myohemoglobinuric ARF. Renal cortical TLR4 protein (Western blotting, immunohistochemistry) and TLR4 mRNA levels (RT-PCR) were determined thereafter (90 min-4 days). Urinary TLR4 excretion post-I/R or CP injection was also assessed. To gain proximal tubule-specific results, TLR4 protein and mRNA were quantified in posthypoxic or oxidant (Fe)-challenged isolated mouse tubules. Finally, TLR4 mRNA was determined in antimycin A-injured cultured proximal tubular (HK-2) cells. Acute in vivo renal injury reduced proximal tubule TLR4 content. These changes corresponded with the appearance of TLR4 fragment(s) in urine and a persistent increase in renal cortical TLR4 mRNA. Isolated proximal tubules responded to injury with rapid TLR4 reductions, dramatic extracellular TLR4 release, and increases in TLR4 mRNA. Glycine blocked these processes, implying membrane pore formation was involved. HK-2 cell injury increased TLR4 mRNA, but not protein levels, suggesting intact transcriptional, but not translational, pathways. Diverse forms of acute tubular injury rapidly reduce proximal tubular TLR4 content. Plasma membrane TLR4 release through glycine-suppressible pores, possibly coupled with a translation block, appears to be involved. Rapid postinjury urinary TLR4 excretion suggests its potential utility as a "biomarker" of impending ARF.


Asunto(s)
Hipoxia/patología , Fallo Renal Crónico/inducido químicamente , Fallo Renal Crónico/patología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Antineoplásicos , Western Blotting , Línea Celular , Cisplatino , Glicerol , Hemoglobinuria/inducido químicamente , Hemoglobinuria/patología , Humanos , Inmunohistoquímica , Corteza Renal/metabolismo , Corteza Renal/patología , Túbulos Renales Proximales/patología , Masculino , Ratones , ARN Mensajero/biosíntesis , Receptor Toll-Like 4/efectos de los fármacos
13.
Am J Respir Cell Mol Biol ; 33(3): 271-9, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15947421

RESUMEN

Bleomycin-induced lung injury triggers a profound and durable increase in tissue inhibitor of metalloproteinase (TIMP)-1 expression, suggesting a potential role for this antiproteinase in the regulation of lung inflammation and fibrosis. TIMP-1 protein induction is spatially restricted to areas of lung injury as determined by immunohistochemistry. Using TIMP-1 null mutation mice, we demonstrate that TIMP-1 deficiency amplifies acute lung injury as determined by exaggerated pulmonary neutrophilia, hemorrhage, and vascular permeability compared with wild-type littermates after bleomycin exposure. The augmented pulmonary neutrophilia observed in TIMP-1-deficient animals was not found in similarly treated TIMP-2-deficient mice. Using TIMP-1 bone marrow (BM) chimeric mice, we observed that the TIMP-1-deficient phenotype was abolished in wild-type recipients of TIMP-1-deficient BM but not in TIMP-1-deficient recipients of wild-type BM. Acute lung injury in TIMP-1-deficient mice was accompanied by exaggerated gelatinase-B activity in the alveolar compartment. TIMP-1 deficiency did not alter neutrophil chemotactic factor accumulation in the injured lung nor neutrophil migration in response to chemotactic stimuli in vivo or in vitro. Moreover, TIMP-1 deficiency did not modify collagen accumulation after bleomycin injury. Our results provide direct evidence that TIMP-1 contributes significantly to the regulation of acute lung injury, functioning to limit inflammation and lung permeability.


Asunto(s)
Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/fisiopatología , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/inmunología , Animales , Antibióticos Antineoplásicos , Bleomicina , Permeabilidad Capilar/inmunología , Quimiotaxis de Leucocito/inmunología , Expresión Génica/inmunología , Hemorragia/inmunología , Hemorragia/mortalidad , Hemorragia/fisiopatología , Lipopolisacáridos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neutrófilos/citología , Neutrófilos/inmunología , Alveolos Pulmonares/enzimología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/patología , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/mortalidad , Fibrosis Pulmonar/fisiopatología , Síndrome de Dificultad Respiratoria/mortalidad , Organismos Libres de Patógenos Específicos , Pérdida de Peso
14.
Blood ; 105(6): 2410-4, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15479725

RESUMEN

T cells recognizing self-peptides are typically deleted in the thymus by negative selection. It is not known whether T cells against persistent viruses (eg, herpesviruses) are generated by the thymus (de novo) after the onset of the infection. Peptides from such viruses might be considered by the thymus as self-peptides, and T cells specific for these peptides might be deleted (negatively selected). Here we demonstrate in baboons infected with baboon cytomegalovirus and baboon lymphocryptovirus (Epstein-Barr virus-like virus) that after autologous transplantation of yellow fluorescent protein (YFP)-marked hematopoietic cells, YFP+ CD4 T cells against these viruses were generated de novo. Thus the thymus generates CD4 T cells against not only pathogens absent from the host but also pathogens present in the host. This finding provides a strong rationale to improve thymopoiesis in recipients of hematopoietic cell transplants and, perhaps, in other persons lacking de novo-generated CD4 T cells, such as AIDS patients and elderly persons.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Células Madre Hematopoyéticas , Linfopoyesis/inmunología , Recuperación de la Función/inmunología , Síndrome de Inmunodeficiencia Adquirida/inmunología , Síndrome de Inmunodeficiencia Adquirida/terapia , Factores de Edad , Animales , Autoantígenos/inmunología , Linfocitos T CD4-Positivos/virología , Citomegalovirus/inmunología , Herpesvirus Humano 4/inmunología , Humanos , Tolerancia Inmunológica , Papio anubis , Papio cynocephalus , Trasplante Autólogo
15.
J Clin Virol ; 25 Suppl 2: S51-6, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12361756

RESUMEN

Cytomegalovirus (CMV) has long been associated with myelosuppression. Evidence for this association has been provided from in vitro studies, statistical analysis of clinical studies, informative case reports, and murine models. Reports differ as to how CMV mediates myelosuppression. Some data indicate direct infection of hematopoietic progenitors and their progeny, others indicate that the supportive microenvironment is infected and thereby its supportive function compromised. In this report we review data suggesting that the severity of myelosuppression in patients is associated with particular CMV genotypes identified by variations in the glycoprotein B gene.


Asunto(s)
Infecciones por Citomegalovirus/fisiopatología , Citomegalovirus/patogenicidad , Células Madre Hematopoyéticas/fisiología , Trasplante de Células Madre/efectos adversos , Animales , Células de la Médula Ósea/fisiología , Células de la Médula Ósea/virología , Células Cultivadas , Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Femenino , Fibroblastos , Variación Genética , Hematopoyesis , Células Madre Hematopoyéticas/virología , Humanos , Ratones , Proteínas del Envoltorio Viral/genética
16.
Cytokine ; 19(1): 37-46, 2002 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-12200112

RESUMEN

We hypothesized that US28, a cytomegalovirus (CMV) CC chemokine receptor homolog, plays a role in modulating the host antiviral defense. Monocyte chemotaxis was induced by supernatants from fibroblasts infected with a US28 deletion mutant of CMV (CMV Delta US28) due to endogenously produced CC chemokines MCP-1 and RANTES. However, these chemokines were sequestered from the supernatants of CMV-infected cells that did express US28. US28 was also capable of sequestering exogenously added RANTES. Surprisingly, cells infected with CMV Delta US28 transcribed and secreted increased levels IL-8, a CXC chemokine, when compared to CMV-infected cells. Finally, because chemokines are potent mediators of immune cell migration through the endothelium, we characterized the CC chemokine binding potential of CMV-infected endothelial cells. We propose that US28 functions as a 'chemokine sink' by sequestering endogenously and exogenously produced chemokines and alters the production of the CXC chemokine IL-8, suggesting that CMV could significantly alter the inflammatory milieu surrounding infected cells.


Asunto(s)
Quimiocinas CC/metabolismo , Citomegalovirus/metabolismo , Interleucina-8/metabolismo , Receptores de Quimiocina/biosíntesis , Proteínas Virales/biosíntesis , Northern Blotting , Células Cultivadas , Quimiocina CCL5 , Quimiocinas/metabolismo , Quimiotaxis , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Ensayo de Inmunoadsorción Enzimática , Humanos , Inflamación , Monocitos/citología , Monocitos/inmunología , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Venas Umbilicales/citología
17.
J Interferon Cytokine Res ; 22(7): 765-72, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12184914

RESUMEN

Previous studies have demonstrated that the intercellular spread of cytomegalovirus (CMV) is reduced in marrow stromal cells that either secrete interleukin-1 (IL-1) or are treated with exogenous IL-1. Here, we report that IL-1-treated marrow stromal cells and fibroblasts, when infected with CMV, produce decreased amounts of infectious progeny virus. CMV-infected cells treated with IL-1 contained more interferon-beta (IFN-beta) mRNA at 24 h postinfection compared with untreated, infected cells. IFN-beta protein secreted into fibroblast culture supernatants increased from 46 +/- 1 IU/ml in untreated, infected cells to 116 +/- 5 IU/ml in IL-1-treated infected cells. When IFN-beta activity was inhibited, using blocking antibodies to either the cytokine or the IFN-alpha/beta receptor, the addition of IL-1 no longer limited viral spread. Furthermore, viral spread in nonIL-1-treated cultures was inhibited by the addition of recombinant IFN-beta. These studies suggest that IL-1 functions to limit CMV spread by increasing the expression of IFN-beta, which in turn reduces production of infectious virus.


Asunto(s)
Antivirales/farmacología , Citomegalovirus/fisiología , Interferón beta/biosíntesis , Interleucina-1/farmacología , Replicación Viral/efectos de los fármacos , Médula Ósea/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/virología , Humanos , Interferón beta/genética , Interferón beta/fisiología , Interleucina-1/genética , Proteínas Recombinantes/farmacología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...